Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sleep Med ; 113: 49-55, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37984017

RESUMO

INTRODUCTION: The basal forebrain (BF) and the medial septum (MS) respectively drive neuronal activity of cerebral cortex and hippocampus (HPC) in sleep-wake cycle. Our previous studies of lesions and neuronal circuit tracing have shown that the pontine parabrachial nucleus (PB) projections to the BF and MS may be a key circuit for cortical and HPC arousal. AIMS: This study aims to demonstrate that PB projections to the BF and MS activate the cerebral cortex and HPC. RESULTS: By using chemogenetic stimulation of the BF, the PB-BF and the PB-MS pathway combined with electroencephalogram (EEG) Fast Fourier Transformation (FFT) analysis in rats, we demonstrated that chemogenetic stimulation of the BF or PB neurons projecting to the BF activated the cerebral cortex while chemogenetic stimulation of the MS or PB neurons projecting to the MS activated HPC activity, in sleep and wake state. These stimulations did not significantly alter sleep-wake amounts. CONCLUSIONS: Our results support that PB projections to the BF and MS specifically regulating cortical and HPC activity.


Assuntos
Prosencéfalo Basal , Núcleos Parabraquiais , Ratos , Animais , Vigília/fisiologia , Prosencéfalo Basal/fisiologia , Nível de Alerta/fisiologia , Eletroencefalografia , Hipocampo
2.
Curr Neurol Neurosci Rep ; 23(12): 907-923, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38060134

RESUMO

PURPOSE OF REVIEW: Since the formal discovery of rapid eye movement (REM) sleep in 1953, we have gained a vast amount of knowledge regarding the specific populations of neurons, their connections, and synaptic mechanisms regulating this stage of sleep and its accompanying features. This article discusses REM sleep circuits and their dysfunction, specifically emphasizing recent studies using conditional genetic tools. RECENT FINDINGS: Sublaterodorsal nucleus (SLD) in the dorsolateral pons, especially the glutamatergic subpopulation in this region (SLDGlut), are shown to be indispensable for REM sleep. These neurons appear to be single REM generators in the rodent brain and may initiate and orchestrate all REM sleep events, including cortical and hippocampal activation and muscle atonia through distinct pathways. However, several cell groups in the brainstem and hypothalamus may influence SLDGlut neuron activity, thereby modulating REM sleep timing, amounts, and architecture. Damage to SLDGlut neurons or their projections involved in muscle atonia leads to REM behavior disorder, whereas the abnormal activation of this pathway during wakefulness may underlie cataplexy in narcolepsy. Despite some opposing views, it has become evident that SLDGlut neurons are the sole generators of REM sleep and its associated characteristics. Further research should prioritize a deeper understanding of their cellular, synaptic, and molecular properties, as well as the mechanisms that trigger their activation during cataplexy and make them susceptible in RBD.


Assuntos
Cataplexia , Narcolepsia , Transtorno do Comportamento do Sono REM , Humanos , Sono REM/fisiologia , Encéfalo
3.
Aging Brain ; 3: 100068, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36911260

RESUMO

Sleep behavior undergoes significant changes across the lifespan, and aging is associated with marked alterations in sleep amounts and quality. The primary sleep changes in healthy older adults include a shift in sleep timing, reduced slow-wave sleep, and impaired sleep maintenance. However, neurodegenerative and psychiatric disorders are more common among the elderly, which further worsen their sleep health. Irrespective of the cause, insufficient sleep adversely affects various bodily functions including energy metabolism, mood, and cognition. In this review, we will focus on the cognitive changes associated with inadequate sleep during normal aging and the underlying neural mechanisms.

4.
CNS Neurosci Ther ; 29(6): 1602-1614, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36794544

RESUMO

AIMS: We often experience dreams of strong irrational and negative emotional contents with postural muscle paralysis during rapid eye movement (REM) sleep, but how REM sleep is generated and its function remain unclear. In this study, we investigate whether the dorsal pontine sub-laterodorsal tegmental nucleus (SLD) is necessary and sufficient for REM sleep and whether REM sleep elimination alters fear memory. METHODS: To investigate whether activation of SLD neurons is sufficient for REM sleep induction, we expressed channelrhodopsin-2 (ChR2) in SLD neurons by bilaterally injecting AAV1-hSyn-ChR2-YFP in rats. We next selectively ablated either glutamatergic or GABAergic neurons from the SLD in mice in order to identify the neuronal subset crucial for REM sleep. We finally  investigated the role of REM sleep in consolidation of fear memory using rat model with complete SLD lesions. RESULTS: We demonstrate the sufficiency of the SLD for REM sleep by showing that photo-activation of ChR2 transfected SLD neurons selectively promotes transitions from non-REM (NREM) sleep to REM sleep in rats. Diphtheria toxin-A (DTA) induced lesions of the SLD in rats or specific deletion of SLD glutamatergic neurons but not GABAergic neurons in mice completely abolish REM sleep, demonstrating the necessity of SLD glutamatergic neurons for REM sleep. We then show that REM sleep elimination by SLD lesions in rats significantly enhances contextual and cued fear memory consolidation by 2.5 and 1.0 folds, respectively, for at least 9 months. Conversely, fear conditioning and fear memory trigger doubled amounts of REM sleep in the following night, and chemo-activation of SLD neurons projecting to the medial septum (MS) selectively enhances hippocampal theta activity in REM sleep; this stimulation immediately after fear acquisition reduces contextual and cued fear memory consolidation by 60% and 30%, respectively. CONCLUSION: SLD glutamatergic neurons generate REM sleep and REM sleep and SLD via the hippocampus particularly down-regulate contextual fear memory.


Assuntos
Medo , Sono REM , Ratos , Camundongos , Animais , Sono REM/fisiologia , Medo/fisiologia , Emoções/fisiologia , Hipocampo , Neurônios GABAérgicos
5.
CNS Neurosci Ther ; 29(4): 1192-1196, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36585816

RESUMO

Pontine sub-laterodorsal tegmental nucleus (SLD) is crucial for REM sleep. However, the necessary role of SLD for REM sleep, cataplexy that resembles REM sleep, and emotion memory by REM sleep has remained unclear. To address these questions, we focally ablated SLD neurons using adenoviral diphtheria-toxin (DTA) approach and found that SLD lesions completely eliminated REM sleep accompanied by wake increase, significantly reduced baseline cataplexy amounts by 40% and reward (sucrose) induced cataplexy amounts by 70% and altered cataplexy EEG Fast Fourier Transform (FFT) from REM sleep-like to wake-like in orexin null (OXKO) mice. We then used OXKO animals with absence of REM sleep and OXKO controls and examined elimination of REM sleep in anxiety and fear extinction. Our resulted showed that REM sleep elimination significantly increased anxiety-like behaviors in open field test (OFT), elevated plus maze test (EPM) and defensive aggression and impaired fear extinction. The data indicate that in OXKO mice the SLD is the sole generator for REM sleep; (2) the SLD selectively mediates REM sleep cataplexy (R-cataplexy) that merges with wake cataplexy (W-cataplexy); (3) REM sleep enhances positive emotion (sucrose induced cataplexy) response, reduces negative emotion state (anxiety), and promotes fear extinction.


Assuntos
Cataplexia , Sono REM , Camundongos , Animais , Sono REM/fisiologia , Cataplexia/genética , Extinção Psicológica , Medo , Emoções
6.
Metabolism ; 130: 155158, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35150732

RESUMO

BACKGROUND: Nearly 14% of Americans experience chronic circadian disruption due to shift work, increasing their risk of obesity, diabetes, and other cardiometabolic disorders. These disorders are also exacerbated by modern eating habits such as frequent snacking and consumption of high-fat foods. METHODS: We investigated the effects of recurrent circadian disruption (RCD) on glucose metabolism in C57BL/6 mice and in human participants exposed to non-24-h light-dark (LD) schedules vs. those on standard 24-h LD schedules. These LD schedules were designed to induce circadian misalignment between behaviors including rest/activity and fasting/eating with the output of the near-24-h central circadian pacemaker, while minimizing sleep loss, and were maintained for 12 weeks in mice and 3 weeks in humans. We examined interactions of these circadian-disrupted schedules compared to control 24-h schedules with a lower-fat diet (LFD, 13% in mouse and 25-27% in humans) and high-fat diet (HFD, 45% in mouse and 45-50% in humans). We also used young vs. older mice to determine whether they would respond differently to RCD. RESULTS: When combined with a HFD, we found that RCD caused significant weight gain in mice and increased body fat in humans, and significantly impaired glucose tolerance and insulin sensitivity in both mice and humans, but this did not occur when RCD was combined with a LFD. This effect was similar in both young and older mice. CONCLUSION: These results in both humans and a model organism indicate that circadian disruption has an adverse effect on metabolism among individuals eating a high-fat Western-style diet, even in the absence of significant sleep loss, and suggest that reducing dietary fat may protect against the metabolic consequences of a lifestyle (such as shift work) that involves chronic circadian disruption.


Assuntos
Dieta Hiperlipídica , Insulina , Animais , Dieta Hiperlipídica/efeitos adversos , Glucose , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia
7.
Curr Neurol Neurosci Rep ; 20(12): 55, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-33006677

RESUMO

PURPOSE OF THE REVIEW: Melanin-concentrating hormone (MCH)-expressing neurons located in the lateral hypothalamus are considered as an integral component of sleep-wake circuitry. However, the precise role of MCH neurons in sleep-wake regulation has remained unclear, despite several years of research employing a wide range of techniques. We review recent data on this aspect, which are mostly inconsistent, and propose a novel role for MCH neurons in sleep regulation. RECENT FINDINGS: While almost all studies using "gain-of-function" approaches show an increase in rapid eye movement sleep (or paradoxical sleep; PS), loss-of-function approaches have not shown reductions in PS. Similarly, the reported changes in wakefulness or non-rapid eye movement sleep (slow-wave sleep; SWS) with manipulation of the MCH system using conditional genetic methods are inconsistent. Currently available data do not support a role for MCH neurons in spontaneous sleep-wake but imply a crucial role for them in orchestrating sleep-wake responses to changes in external and internal environments.


Assuntos
Hormônios Hipotalâmicos , Humanos , Hormônios Hipotalâmicos/genética , Melaninas , Neurônios , Hormônios Hipofisários/genética , Sono , Vigília
8.
J Headache Pain ; 21(1): 86, 2020 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-32631251

RESUMO

BACKGROUND: Migraine is a common headache disorder, with cortical spreading depolarization (CSD) considered as the underlying electrophysiological event. CSD is a slowly propagating wave of neuronal and glial depolarization. Sleep disorders are well known risk factors for migraine chronification, and changes in wake-sleep pattern such as sleep deprivation are common migraine triggers. The underlying mechanisms are unknown. As a step towards developing an animal model to study this, we test whether sleep deprivation, a modifiable migraine trigger, enhances CSD susceptibility in rodent models. METHODS: Acute sleep deprivation was achieved using the "gentle handling method", chosen to minimize stress and avoid confounding bias. Sleep deprivation was started with onset of light (diurnal lighting conditions), and assessment of CSD was performed at the end of a 6 h or 12 h sleep deprivation period. The effect of chronic sleep deprivation on CSD was assessed 6 weeks or 12 weeks after lesioning of the hypothalamic ventrolateral preoptic nucleus. All experiments were done in a blinded fashion with respect to sleep status. During 60 min of continuous topical KCl application, we assessed the total number of CSDs, the direct current shift amplitude and duration of the first CSD, the average and cumulative duration of all CSDs, propagation speed, and electrical CSD threshold. RESULTS: Acute sleep deprivation of 6 h (n = 17) or 12 h (n = 11) duration significantly increased CSD frequency compared to controls (17 ± 4 and 18 ± 2, respectively, vs. 14 ± 2 CSDs/hour in controls; p = 0.003 for both), whereas other electrophysiological properties of CSD were unchanged. Acute total sleep deprivation over 12 h but not over 6 h reduced the electrical threshold of CSD compared to controls (p = 0.037 and p = 0.095, respectively). Chronic partial sleep deprivation in contrast did not affect CSD susceptibility in rats. CONCLUSIONS: Acute but not chronic sleep deprivation enhances CSD susceptibility in rodents, possibly underlying its negative impact as a migraine trigger and exacerbating factor. Our findings underscore the importance of CSD as a therapeutic target in migraine and suggest that headache management should identify and treat associated sleep disorders.


Assuntos
Enxaqueca sem Aura/fisiopatologia , Privação do Sono/fisiopatologia , Animais , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley
9.
Eur J Neurosci ; 52(9): 4100-4114, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32588491

RESUMO

Sleep pressure that builds up gradually during the extended wakefulness results in sleep rebound. Several lines of evidence, however, suggest that wake per se may not be sufficient to drive sleep rebound and that rapid eye movement (REM) and non-rapid eye movement (NREM) sleep rebound may be differentially regulated. In this study, we investigated the relative contribution of brain versus physical activities in REM and NREM sleep rebound by four sets of experiments. First, we forced locomotion in rats in a rotating wheel for 4 hr and examined subsequent sleep rebound. Second, we exposed the rats lacking homeostatic sleep response after prolonged quiet wakefulness and arousal brain activity induced by chemoactivation of parabrachial nucleus to the same rotating wheel paradigm and tested if physical activity could rescue the sleep homeostasis. Third, we varied motor activity levels while concurrently inhibiting the cortical activity by administering ketamine or xylazine (motor inhibitor), or ketamine + xylazine mixture and investigated if motor activity in the absence of activated cortex can cause NREM sleep rebound. Fourth and finally, we manipulated cortical activity by administering ketamine (that induced active wakefulness and waking brain) alone or in combination with atropine (that selectively inhibits the cortex) and studied if cortical inhibition irrespective of motor activity levels can block REM sleep rebound. Our results demonstrate that motor activity but not cortical activity determines NREM sleep rebound whereas cortical activity but not motor activity determines REM sleep rebound.


Assuntos
Eletroencefalografia , Sono , Animais , Homeostase , Ratos , Sono REM , Vigília
10.
Eur J Neurosci ; 51(12): 2343-2354, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-30269396

RESUMO

A single phase advance of the light:dark (LD) cycle can temporarily disrupt synchrony of neural circadian rhythms within the suprachiasmatic nucleus (SCN) and between the SCN and peripheral tissues. Compounding this, modern life can involve repeated disruptive light conditions. To model chronic disruption to the circadian system, we exposed male mice to more than a month of a 20-hr light cycle (LD10:10), which mice typically cannot entrain to. Control animals were housed under LD12:12. We measured locomotor activity and body temperature rhythms in vivo, and rhythms of PER2::LUC bioluminescence in SCN and peripheral tissues ex vivo. Unexpectedly, we discovered strong effects of the time of dissection on circadian phase of PER2::LUC bioluminescent rhythms, which varied across tissues. White adipose tissue was strongly reset by dissection, while thymus phase appeared independent of dissection timing. Prior light exposure impacted the SCN, resulting in strong resetting of SCN phase by dissection for mice housed under LD10:10, and weak phase shifts by time of dissection in SCN from control LD12:12 mice. These findings suggest that exposure to circadian disruption may desynchronize SCN neurons, increasing network sensitivity to perturbations. We propose that tissues with a weakened circadian network, such as the SCN under disruptive light conditions, or with little to no coupling, for example, some peripheral tissues, will show increased resetting effects. In particular, exposure to light at inconsistent circadian times on a recurring weekly basis disrupts circadian rhythms and alters sensitivity of the SCN neural pacemaker to dissection time.


Assuntos
Relógios Circadianos , Animais , Ritmo Circadiano , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Núcleo Supraquiasmático/metabolismo
11.
J Neurosci ; 40(1): 171-190, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31694962

RESUMO

Origin and functions of intermittent transitions among sleep stages, including brief awakenings and arousals, constitute a challenge to the current homeostatic framework for sleep regulation, focusing on factors modulating sleep over large time scales. Here we propose that the complex micro-architecture characterizing sleep on scales of seconds and minutes results from intrinsic non-equilibrium critical dynamics. We investigate θ- and δ-wave dynamics in control rats and in rats where the sleep-promoting ventrolateral preoptic nucleus (VLPO) is lesioned (male Sprague-Dawley rats). We demonstrate that bursts in θ and δ cortical rhythms exhibit complex temporal organization, with long-range correlations and robust duality of power-law (θ-bursts, active phase) and exponential-like (δ-bursts, quiescent phase) duration distributions, features typical of non-equilibrium systems self-organizing at criticality. We show that such non-equilibrium behavior relates to anti-correlated coupling between θ- and δ-bursts, persists across a range of time scales, and is independent of the dominant physiologic state; indications of a basic principle in sleep regulation. Further, we find that VLPO lesions lead to a modulation of cortical dynamics resulting in altered dynamical parameters of θ- and δ-bursts and significant reduction in θ-δ coupling. Our empirical findings and model simulations demonstrate that θ-δ coupling is essential for the emerging non-equilibrium critical dynamics observed across the sleep-wake cycle, and indicate that VLPO neurons may have dual role for both sleep and arousal/brief wake activation. The uncovered critical behavior in sleep- and wake-related cortical rhythms indicates a mechanism essential for the micro-architecture of spontaneous sleep-stage and arousal transitions within a novel, non-homeostatic paradigm of sleep regulation.SIGNIFICANCE STATEMENT We show that the complex micro-architecture of sleep-stage/arousal transitions arises from intrinsic non-equilibrium critical dynamics, connecting the temporal organization of dominant cortical rhythms with empirical observations across scales. We link such behavior to sleep-promoting neuronal population, and demonstrate that VLPO lesion (model of insomnia) alters dynamical features of θ and δ rhythms, and leads to significant reduction in θ-δ coupling. This indicates that VLPO neurons may have dual role for both sleep and arousal/brief wake control. The reported empirical findings and modeling simulations constitute first evidences of a neurophysiological fingerprint of self-organization and criticality in sleep- and wake-related cortical rhythms; a mechanism essential for spontaneous sleep-stage and arousal transitions that lays the bases for a novel, non-homeostatic paradigm of sleep regulation.


Assuntos
Sono/fisiologia , Vigília/fisiologia , Animais , Ritmo Delta , Eletroencefalografia , Masculino , Área Pré-Óptica/lesões , Área Pré-Óptica/fisiologia , Ratos , Ratos Sprague-Dawley , Fases do Sono/fisiologia , Organismos Livres de Patógenos Específicos , Ritmo Teta
12.
Neuroscience ; 406: 314-324, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30890480

RESUMO

Neurons containing melanin-concentrating hormone (MCH) in the lateral hypothalamic area (LH) have been shown to promote rapid eye movement sleep (REMs) in mice. However, the downstream neural pathways through which MCH neurons influence REMs remained unclear. Because MCH neurons are considered to be primarily inhibitory, we hypothesized that these neurons inhibit the midbrain 'REMs-suppressing' region consisting of the ventrolateral periaqueductal gray and the lateral pontine tegmentum (vlPAG/LPT) to promote REMs. To test this hypothesis, we optogenetically inhibited MCH terminals in the vlPAG/LPT under baseline conditions as well as with simultaneous chemogenetic activation of MCH soma. We found that inhibition of MCH terminals in the vlPAG/LPT significantly reduced transitions into REMs during spontaneous sleep-wake cycles and prevented the increase in REMs transitions observed after chemogenetic activation of MCH neurons. These results strongly suggest that the vlPAG/LPT may be an essential relay through which MCH neurons modulate REMs.


Assuntos
Movimentos Oculares/fisiologia , Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Substância Cinzenta Periaquedutal/fisiologia , Hormônios Hipofisários/metabolismo , Sono REM/fisiologia , Animais , Região Hipotalâmica Lateral/fisiologia , Masculino , Camundongos Transgênicos , Vias Neurais/fisiologia , Neurônios/fisiologia , Vigília/fisiologia
13.
PLoS Biol ; 17(3): e3000172, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30893297

RESUMO

Sleep and wakefulness are greatly influenced by various physiological and psychological factors, but the neuronal elements responsible for organizing sleep-wake behavior in response to these factors are largely unknown. In this study, we report that a subset of neurons in the lateral hypothalamic area (LH) expressing the neuropeptide neurotensin (Nts) is critical for orchestrating sleep-wake responses to acute psychological and physiological challenges or stressors. We show that selective activation of NtsLH neurons with chemogenetic or optogenetic methods elicits rapid transitions from non-rapid eye movement (NREM) sleep to wakefulness and produces sustained arousal, higher locomotor activity (LMA), and hyperthermia, which are commonly observed after acute stress exposure. On the other hand, selective chemogenetic inhibition of NtsLH neurons attenuates the arousal, LMA, and body temperature (Tb) responses to a psychological stress (a novel environment) and augments the responses to a physiological stress (fasting).


Assuntos
Febre/metabolismo , Região Hipotalâmica Lateral/metabolismo , Neurotensina/metabolismo , Animais , Temperatura Corporal , Eletroforese , Técnicas de Genotipagem , Locomoção/fisiologia , Masculino , Camundongos , Neurônios/metabolismo
14.
Brain Struct Funct ; 224(1): 99-110, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30284033

RESUMO

Neurons containing melanin-concentrating hormone (MCH) in the posterior lateral hypothalamus play an integral role in rapid eye movement sleep (REMs) regulation. As MCH neurons also contain a variety of other neuropeptides [e.g., cocaine- and amphetamine-regulated transcript (CART) and nesfatin-1] and neurotransmitters (e.g., glutamate), the specific neurotransmitter responsible for REMs regulation is not known. We hypothesized that glutamate, the primary fast-acting neurotransmitter in MCH neurons, is necessary for REMs regulation. To test this hypothesis, we deleted vesicular glutamate transporter (Vglut2; necessary for synaptic release of glutamate) specifically from MCH neurons by crossing MCH-Cre mice (expressing Cre recombinase in MCH neurons) with Vglut2flox/flox mice (expressing LoxP-modified alleles of Vglut2), and studied the amounts, architecture and diurnal variation of sleep-wake states during baseline conditions. We then activated the MCH neurons lacking glutamate neurotransmission using chemogenetic methods and tested whether these MCH neurons still promoted REMs. Our results indicate that glutamate in MCH neurons contributes to normal diurnal variability of REMs by regulating the levels of REMs during the dark period, but MCH neurons can promote REMs even in the absence of glutamate.


Assuntos
Ritmo Circadiano , Ácido Glutâmico/metabolismo , Hormônios Hipotalâmicos/metabolismo , Hipotálamo Posterior/metabolismo , Melaninas/metabolismo , Neurônios/metabolismo , Hormônios Hipofisários/metabolismo , Sono REM , Animais , Hormônios Hipotalâmicos/genética , Hipotálamo Posterior/citologia , Masculino , Melaninas/genética , Camundongos Transgênicos , Fotoperíodo , Hormônios Hipofisários/genética , Fatores de Tempo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Vigília
15.
Nat Commun ; 9(1): 4129, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30297727

RESUMO

The preoptic area (POA) is necessary for sleep, but the fundamental POA circuits have remained elusive. Previous studies showed that galanin (GAL)- and GABA-producing neurons in the ventrolateral preoptic nucleus (VLPO) express cFos after periods of increased sleep and innervate key wake-promoting regions. Although lesions in this region can produce insomnia, high frequency photostimulation of the POAGAL neurons was shown to paradoxically cause waking, not sleep. Here we report that photostimulation of VLPOGAL neurons in mice promotes sleep with low frequency stimulation (1-4 Hz), but causes conduction block and waking at frequencies above 8 Hz. Further, optogenetic inhibition reduces sleep. Chemogenetic activation of VLPOGAL neurons confirms the increase in sleep, and also reduces body temperature. In addition, chemogenetic activation of VLPOGAL neurons induces short-latency sleep in an animal model of insomnia. Collectively, these findings establish a causal role of VLPOGAL neurons in both sleep induction and heat loss.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Galanina/metabolismo , Neurônios/metabolismo , Área Pré-Óptica/metabolismo , Sono/fisiologia , Animais , Regulação da Temperatura Corporal/genética , Eletroencefalografia , Eletromiografia , Galanina/genética , Perfilação da Expressão Gênica , Masculino , Camundongos Transgênicos , Área Pré-Óptica/citologia , Sono/genética , Distúrbios do Início e da Manutenção do Sono/genética , Distúrbios do Início e da Manutenção do Sono/metabolismo , Distúrbios do Início e da Manutenção do Sono/fisiopatologia
16.
Neurobiol Dis ; 120: 12-20, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30149182

RESUMO

The lateral hypothalamus contains neurons producing orexins that promote wakefulness and suppress REM sleep as well as neurons producing melanin-concentrating hormone (MCH) that likely promote REM sleep. Narcolepsy with cataplexy is caused by selective loss of the orexin neurons, and the MCH neurons appear unaffected. As the orexin and MCH systems exert opposing effects on REM sleep, we hypothesized that imbalance in this REM sleep-regulating system due to activity in the MCH neurons may contribute to the striking REM sleep dysfunction characteristic of narcolepsy. To test this hypothesis, we chemogenetically activated the MCH neurons and pharmacologically blocked MCH signaling in a murine model of narcolepsy and studied the effects on sleep-wake behavior and cataplexy. To chemoactivate MCH neurons, we injected an adeno-associated viral vector containing the hM3Dq stimulatory DREADD into the lateral hypothalamus of orexin null mice that also express Cre recombinase in the MCH neurons (MCH-Cre::OX-KO mice) and into control MCH-Cre mice with normal orexin expression. In both lines of mice, activation of MCH neurons by clozapine-N-oxide (CNO) increased rapid eye movement (REM) sleep without altering other states. In mice lacking orexins, activation of the MCH neurons also increased abnormal intrusions of REM sleep manifest as cataplexy and short latency transitions into REM sleep (SLREM). Conversely, a MCH receptor 1 antagonist, SNAP 94847, almost completely eliminated SLREM and cataplexy in OX-KO mice. These findings affirm that MCH neurons promote REM sleep under normal circumstances, and their activity in mice lacking orexins likely triggers abnormal intrusions of REM sleep into non-REM sleep and wake, resulting in the SLREM and cataplexy characteristic of narcolepsy.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Narcolepsia/metabolismo , Neurônios/fisiologia , Hormônios Hipofisários/metabolismo , Sono REM/fisiologia , Animais , Feminino , Hormônios Hipotalâmicos/antagonistas & inibidores , Hormônios Hipotalâmicos/genética , Masculino , Melaninas/antagonistas & inibidores , Melaninas/genética , Camundongos , Camundongos Knockout , Narcolepsia/genética , Neurônios/efeitos dos fármacos , Piperidinas/farmacologia , Hormônios Hipofisários/antagonistas & inibidores , Hormônios Hipofisários/genética
17.
Exp Neurol ; 290: 53-62, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28077261

RESUMO

Discrete populations of neurons at multiple levels of the brainstem control rapid eye movement (REM) sleep and the accompanying loss of postural muscle tone, or atonia. The specific contributions of these brainstem cell populations to REM sleep control remains incompletely understood. Here we show in rats that viral vector-based lesions of the ventromedial medulla at a level rostral to the inferior olive (pSOM) produced violent myoclonic twitches and abnormal electromyographic spikes, but not complete loss of tonic atonia, during REM sleep. Motor tone during non-REM (NREM) sleep was unaffected in these same animals. Acute chemogenetic activation of pSOM neurons in rats robustly and selectively suppressed REM sleep but not NREM sleep. Similar lesions targeting the more rostral ventromedial medulla (RVM) did not affect sleep or atonia, while chemogenetic stimulation of the RVM produced wakefulness and reduced sleep. Finally, selective activation of vesicular GABA transporter (VGAT) pSOM neurons in mice produced complete suppression of REM sleep whereas their loss increased EMG spikes during REM sleep. These results reveal a key contribution of the pSOM and specifically the VGAT+ neurons in this region in REM sleep and motor control.


Assuntos
Bulbo/fisiologia , Sono REM/fisiologia , Adenoviridae/genética , Animais , Eletroencefalografia , Eletromiografia , Imuno-Histoquímica , Masculino , Camundongos , Mioclonia/fisiopatologia , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Vigília/efeitos dos fármacos
18.
Neuroscience ; 336: 102-113, 2016 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-27595887

RESUMO

Currently available evidence indicates that neurons containing melanin-concentrating hormone (MCH) in the lateral hypothalamus are critical modulators of sleep-wakefulness, but their precise role in this function is not clear. Studies employing optogenetic stimulation of MCH neurons have yielded inconsistent results, presumably due to differences in the optogenetic stimulation protocols, which do not approximate normal patterns of cell firing. In order to resolve this discrepancy, we (1) selectively activated the MCH neurons using a chemogenetic approach (Cre-dependent hM3Dq expression) and (2) selectively destroyed MCH neurons using a genetically targeted diphtheria toxin deletion method, and studied the changes in sleep-wake in mice. Our results indicate that selective activation of MCH neurons causes specific increases in rapid eye movement (REM) sleep without altering wake or non-REM (NREM) sleep. On the other hand, selective deletions of MCH neurons altered the diurnal rhythm of wake and REM sleep without altering their total amounts. These results indicate that activation of MCH neurons primarily drives REM sleep and their presence may be necessary for normal expression of diurnal variation of REM sleep and wake.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Neurônios/metabolismo , Hormônios Hipofisários/metabolismo , Sono REM/fisiologia , Animais , Comportamento Animal/fisiologia , Camundongos Transgênicos , Optogenética/métodos , Vigília
20.
Nature ; 532(7597): 112-6, 2016 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-27027295

RESUMO

Brown and beige adipose tissues can dissipate chemical energy as heat through thermogenic respiration, which requires uncoupling protein 1 (UCP1). Thermogenesis from these adipocytes can combat obesity and diabetes, encouraging investigation of factors that control UCP1-dependent respiration in vivo. Here we show that acutely activated thermogenesis in brown adipose tissue is defined by a substantial increase in levels of mitochondrial reactive oxygen species (ROS). Remarkably, this process supports in vivo thermogenesis, as pharmacological depletion of mitochondrial ROS results in hypothermia upon cold exposure, and inhibits UCP1-dependent increases in whole-body energy expenditure. We further establish that thermogenic ROS alter the redox status of cysteine thiols in brown adipose tissue to drive increased respiration, and that Cys253 of UCP1 is a key target. UCP1 Cys253 is sulfenylated during thermogenesis, while mutation of this site desensitizes the purine-nucleotide-inhibited state of the carrier to adrenergic activation and uncoupling. These studies identify mitochondrial ROS induction in brown adipose tissue as a mechanism that supports UCP1-dependent thermogenesis and whole-body energy expenditure, which opens the way to improved therapeutic strategies for combating metabolic disorders.


Assuntos
Cisteína/química , Metabolismo Energético , Canais Iônicos/química , Canais Iônicos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Termogênese , Tecido Adiposo Marrom/química , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/metabolismo , Animais , Respiração Celular , Cisteína/genética , Cisteína/metabolismo , Metabolismo Energético/efeitos dos fármacos , Feminino , Humanos , Canais Iônicos/deficiência , Canais Iônicos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Oxirredução , Compostos de Sulfidrila/metabolismo , Termogênese/efeitos dos fármacos , Proteína Desacopladora 1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...